Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 378
Filter
1.
J Intern Med ; 283(1): 16-44, 2018 01.
Article in English | MEDLINE | ID: mdl-28940884

ABSTRACT

Platelet-derived growth factor (PDGF) isoforms and their receptors have important roles during embryogenesis, particularly in the development of various mesenchymal cell types in different organs. In the adult, PDGF stimulates wound healing and regulates tissue homeostasis. However, overactivity of PDGF signalling is associated with malignancies and other diseases characterized by excessive cell proliferation, such as fibrotic conditions and atherosclerosis. In certain tumours, genetic or epigenetic alterations of the genes for PDGF ligands and receptors drive tumour cell proliferation and survival. Examples include the rare skin tumour dermatofibrosarcoma protuberance, which is driven by autocrine PDGF stimulation due to translocation of a PDGF gene, and certain gastrointestinal stromal tumours and leukaemias, which are driven by constitute activation of PDGF receptors due to point mutations and formation of fusion proteins of the receptors, respectively. Moreover, PDGF stimulates cells in tumour stroma and promotes angiogenesis as well as the development of cancer-associated fibroblasts, both of which promote tumour progression. Inhibitors of PDGF signalling may thus be of clinical usefulness in the treatment of certain tumours.


Subject(s)
Carcinogenesis , Neoplasms , Neovascularization, Pathologic/metabolism , Platelet-Derived Growth Factor , Receptors, Platelet-Derived Growth Factor , Animals , Antineoplastic Agents/pharmacology , Carcinogenesis/genetics , Carcinogenesis/metabolism , Humans , Ligands , Neoplasms/drug therapy , Neoplasms/genetics , Neoplasms/metabolism , Platelet-Derived Growth Factor/antagonists & inhibitors , Platelet-Derived Growth Factor/genetics , Platelet-Derived Growth Factor/metabolism , Protein Kinase Inhibitors/pharmacology , Receptors, Platelet-Derived Growth Factor/antagonists & inhibitors , Receptors, Platelet-Derived Growth Factor/genetics , Receptors, Platelet-Derived Growth Factor/metabolism , Signal Transduction
2.
Oncogenesis ; 6(6): e348, 2017 Jun 12.
Article in English | MEDLINE | ID: mdl-28604766

ABSTRACT

The levels of hyaluronan, a ubiquitous glycosaminoglycan prominent in the extracellular matrix, is balanced through the actions of hyaluronan-synthesizing enzymes (HAS1, 2 and 3) and degrading hyaluronidases (Hyal 1, 2, 3 and PH20). Hyaluronan accumulates in rapidly remodeling tissues, such as breast cancer, due to deregulated expression of the HAS2 gene and/or alterations of HAS2 activity. The activity of HAS2 is regulated by post-translational modifications, including ubiquitination. In order to identify deubiquitinating enzymes (DUBs) that are involved in de-ubiquitination of HAS2, a complementary (cDNA) library of 69 Flag-HA-tagged human DUBs cloned into retroviral vectors was screened in human embryonic kidney (HEK) 293T cells for their ability to de-ubiquitinate myc-tagged HAS2. Several DUBs were found to decrease the ubiquitination of 6myc-HAS2, among which, the most effective were USP17 and USP4. USP17 efficiently removed polyubiquitination, whereas USP4 preferentially removed monoubiquitination of 6myc-HAS2. Co-immunoprecipitation studies revealed interactions between HAS2 and USP17, as well as between HAS2 and USP4, in membrane preparations of HEK293T cells. USP17 significantly stabilized 6myc-HAS2 protein levels, whereas USP4 did not. The silencing of USP17 led to decreased hyaluronan production, whereas the suppression of USP4 increased hyaluronan synthesis. Importantly, high levels of USP17 and HAS2 were detected in a panel of cancer cell lines compared to normal cells, and immunohistochemical stainings revealed higher expression of USP17 and HAS2 in tissues of lung cancer patients compared to normal tissue. In conclusion, USP17 and USP4 differently affect HAS2 ubiquitination, and the stability and function of HAS2.

3.
Oncogene ; 32(47): 5409-20, 2013 Nov 21.
Article in English | MEDLINE | ID: mdl-23524585

ABSTRACT

Glioblastoma multiforme (GBM) is an aggressive brain malignancy characterized by high heterogeneity and invasiveness. It is increasingly accepted that the refractory feature of GBM to current therapies stems from the existence of few tumorigenic cells that sustain tumor growth and spreading, the so-called glioma-initiating cells (GICs). Previous studies showed that cytokines of the bone morphogenetic protein (BMP) family induce differentiation of the GICs, and thus act as tumor suppressors. Molecular pathways that explain this behavior of BMP cytokines remain largely elusive. Here, we show that BMP signaling induces Smad-dependent expression of the transcriptional regulator Snail in a rapid and sustained manner. Consistent with its already established promigratory function in other cell types, we report that Snail silencing decreases GBM cell migration. Consequently, overexpression of Snail increases GBM invasiveness in a mouse xenograft model. Surprisingly, we found that Snail depletes the GBM capacity to form gliomaspheres in vitro and to grow tumors in vivo, both of which are important features shared by GICs. Thus Snail, acting downstream of BMP signaling, dissociates the invasive capacity of GBM cells from their tumorigenic potential.


Subject(s)
Brain Neoplasms/pathology , Glioblastoma/pathology , Transcription Factors/metabolism , Animals , Bone Morphogenetic Proteins/metabolism , Brain Neoplasms/genetics , Brain Neoplasms/metabolism , Cell Differentiation/genetics , Cell Line, Tumor , Cell Movement/genetics , Cell Proliferation , Gene Expression Regulation, Neoplastic , Glioblastoma/genetics , Glioblastoma/metabolism , Humans , Mice , Neoplastic Stem Cells/metabolism , RNA Interference , RNA, Small Interfering , Signal Transduction/genetics , Smad1 Protein/genetics , Smad2 Protein/genetics , Smad3 Protein/genetics , Smad4 Protein/genetics , Smad5 Protein/genetics , Snail Family Transcription Factors , Transcription Factors/genetics
4.
Oncogene ; 32(13): 1609-15, 2013 Mar 28.
Article in English | MEDLINE | ID: mdl-22614010

ABSTRACT

A dual role of transforming growth factor ß (TGF-ß), to both suppress and promote tumor progression and metastasis, has been well established, but its molecular basis has remained elusive. In this review, we focus on Smad proteins, which are central mediators of the signal transduction of TGF-ß family members. We describe current knowledge of cell-type-specific binding patterns of Smad proteins and mechanisms of transcriptional regulation, obtained from recent studies on genome-wide binding sites of Smad molecules. We also discuss potential application of the genome-wide analyses for cancer research, which will allow clarification of the complex mechanisms occurring during cancer progression, and the identification of potential biomarkers for future cancer diagnosis, prognosis and therapy.


Subject(s)
Gene Expression Profiling , Gene Expression Regulation , Smad Proteins/metabolism , Smad Proteins/physiology , Animals , Chromatin Immunoprecipitation/methods , Gene Expression Profiling/methods , Genome/genetics , Genome/physiology , Humans , Models, Biological , Oligonucleotide Array Sequence Analysis/methods , Protein Binding/genetics , Protein Binding/physiology , Smad Proteins/genetics
5.
Oncogene ; 32(37): 4355-65, 2013 Sep 12.
Article in English | MEDLINE | ID: mdl-23108409

ABSTRACT

Epithelial-mesenchymal transition (EMT) is a developmental program, which can be adopted by cancer cells to increase their migration and ability to form metastases. Transforming growth factor ß (TGFß) is a well-studied inducer of EMT. We demonstrate that TGFß potently stimulates hyaluronan synthesis via upregulation of hyaluronan synthase 2 (HAS2) in NMuMG mammary epithelial cells. This stimulatory effect requires the kinase active type I TGFß receptor and is dependent on Smad signaling and activation of the p38 mitogen-activated protein kinase. Knockdown of HAS2 inhibited the TGFß-induced EMT by about 50%, as determined by the phase contrast microscopy and immunostaining using the EMT marker ZO-1. Furthermore, real-time PCR analysis of the EMT markers fibronectin, Snail1 and Zeb1 revealed decreased expressions upon HAS2 suppression, using specific small interfering RNA (siRNA) for HAS2. Removal of the extracellular hyaluronan by Streptomyces hyaluronidase or inhibiting the binding to its cell surface receptor CD44 by blocking antibodies, did not inhibit TGFß-induced EMT. Interestingly, HAS2 suppression completely abolished the TGFß-induced cell migration, whereas CD44 knockdown did not. These observations suggest that TGFß-dependent HAS2 expression, but not extracellular hyaluronan, has an important regulatory role in TGFß-induced EMT.


Subject(s)
Epithelial-Mesenchymal Transition/drug effects , Epithelial-Mesenchymal Transition/genetics , Glucuronosyltransferase/genetics , Transforming Growth Factor beta/pharmacology , Cell Movement/drug effects , Cell Movement/genetics , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Gene Knockdown Techniques , Glucuronosyltransferase/metabolism , Humans , Hyaluronan Receptors/genetics , Hyaluronan Receptors/metabolism , Hyaluronan Synthases , Hyaluronic Acid/biosynthesis , Mammary Glands, Human/metabolism , Mammary Glands, Human/pathology , Protein Serine-Threonine Kinases/metabolism , Receptor, Transforming Growth Factor-beta Type I , Receptors, Transforming Growth Factor beta/metabolism , Signal Transduction , Smad Proteins/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism
6.
Oncogene ; 32(31): 3606-15, 2013 Aug 01.
Article in English | MEDLINE | ID: mdl-22926518

ABSTRACT

Deregulation of the transforming growth factor ß (TGFß) signal transduction cascade is functionally linked to cancer. In early phases, TGFß acts as a tumor suppressor by inhibiting tumor cell proliferation, whereas in late phases, it can act as a tumor promoter by stimulating tumor cell invasion and metastasis. Smad transcriptional effectors mediate TGFß responses, but relatively little is known about the Smad-containing complexes that are important for epithelial-mesenchymal transition and invasion. In this study, we have tested the hypothesis that specific members of the AP-1 transcription factor family determine TGFß signaling specificity in breast cancer cell invasion. Using a 3D model of collagen-embedded spheroids of MCF10A-MII premalignant human breast cancer cells, we identified the AP-1 transcription factor components c-Jun, JunB, c-Fos and Fra1 as essential factors for TGFß-induced invasion and found that various mesenchymal and invasion-associated TGFß-induced genes are co-regulated by these proteins. In situ proximity ligation assays showed that TGFß signaling not only induces complexes between Smad3 and Smad4 in the nucleus but also complexes between Smad2/3 and Fra1, whereas complexes between Smad3, c-Jun and JunB could already be detected before TGFß stimulation. Finally, chromatin immunoprecipitations showed that c-Jun, JunB and Fra1, but not c-Fos, are required for TGFß-induced binding of Smad2/3 to the mmp-10 and pai-1 promoters. Together these results suggest that in particular formation of Smad2/3-Fra1 complexes may reflect activation of the Smad/AP-1-dependent TGFß-induced invasion program.


Subject(s)
Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Smad Proteins/metabolism , Transcription Factor AP-1/metabolism , Transforming Growth Factor beta/pharmacology , Cell Line, Tumor , Enzyme Activation/drug effects , Humans , Matrix Metalloproteinases/genetics , Mesoderm/drug effects , Mesoderm/metabolism , Mesoderm/pathology , Neoplasm Invasiveness , Plasminogen Activator Inhibitor 1/genetics , Promoter Regions, Genetic/genetics , Protein Binding , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins c-fos/metabolism , Proto-Oncogene Proteins c-jun/metabolism , Receptor, Transforming Growth Factor-beta Type I , Receptors, Transforming Growth Factor beta/metabolism , Signal Transduction/drug effects , Spheroids, Cellular/drug effects , Spheroids, Cellular/metabolism , Spheroids, Cellular/pathology , Transcription Factors/metabolism , Transforming Growth Factor beta/metabolism
7.
Mol Biol Cell ; 23(13): 2571-82, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22573884

ABSTRACT

Receptor tyrosine kinase (RTK) signaling is frequently increased in tumor cells, sometimes as a result of decreased receptor down-regulation. The extent to which the endocytic trafficking routes can contribute to such RTK hyperactivation is unclear. Here, we show for the first time that fibroblast transformation by H-RasG12V induces the internalization of platelet-derived growth factor ß-receptor (PDGFRß) by macropinocytosis, enhancing its signaling activity and increasing anchorage-independent proliferation. H-RasG12V transformation and PDGFRß activation were synergistic in stimulating phosphatidylinositol (PI) 3-kinase activity, leading to receptor macropinocytosis. PDGFRß macropinocytosis was both necessary and sufficient for enhanced receptor activation. Blocking macropinocytosis by inhibition of PI 3-kinase prevented the increase in receptor activity in transformed cells. Conversely, increasing macropinocytosis by Rabankyrin-5 overexpression was sufficient to enhance PDGFRß activation in nontransformed cells. Simultaneous stimulation with PDGF-BB and epidermal growth factor promoted macropinocytosis of both receptors and increased their activation in nontransformed cells. We propose that H-Ras transformation promotes tumor progression by enhancing growth factor receptor signaling as a result of increased receptor macropinocytosis.


Subject(s)
Cell Transformation, Neoplastic/genetics , Fibroblasts/metabolism , Pinocytosis , Proto-Oncogene Proteins p21(ras)/genetics , Receptor, Platelet-Derived Growth Factor beta/metabolism , Becaplermin , Cell Adhesion , Cell Proliferation , Cells, Cultured , Epidermal Growth Factor/physiology , ErbB Receptors/metabolism , Fibroblasts/physiology , Humans , Mutation, Missense , Phosphorylation , Protein Processing, Post-Translational , Protein Transport , Proto-Oncogene Proteins c-sis/physiology , Signal Transduction , Transport Vesicles/metabolism
8.
Recent Results Cancer Res ; 180: 103-14, 2010.
Article in English | MEDLINE | ID: mdl-20033380

ABSTRACT

Pericytes are smooth muscle-like cells found in close contact with the endothelium in capillaries, where they regulate the morphology and function of the vessels. During vessel formation, platelet-derived growth factor-BB (PDGF-BB) is required for the recruitment and differentiation of pericytes. Tumor vessels display abnormal morphology and increased endothelial proliferation, resulting in leaky, tortuous vessels that are often poorly perfused. These vessels typically display decreased pericyte density, and the tumor-associated pericytes often express abnormal markers and show abnormal morphology. Anti-angiogenic therapy targeting pro-angiogenic growth factor pathways has been applied to a broad range of solid tumors with varying results. Studies utilizing mouse models indicate that the presence of pericytes protect endothelial cells against inhibition of vascular endothelial growth factor (VEGF) signaling. Simultaneous inhibition of PDGF receptors on pericytes therefore improves the effect of VEGF inhibitors on endothelial cells and enhances anti-angiogenic therapy.


Subject(s)
Neovascularization, Physiologic/physiology , Platelet-Derived Growth Factor/physiology , Angiogenesis Inhibitors/therapeutic use , Animals , Humans , Neoplasms/blood supply , Neoplasms/drug therapy , Pericytes/drug effects , Pericytes/physiology , Receptors, Platelet-Derived Growth Factor/antagonists & inhibitors , Receptors, Platelet-Derived Growth Factor/physiology
9.
Oncogene ; 25(35): 4913-22, 2006 Aug 10.
Article in English | MEDLINE | ID: mdl-16547494

ABSTRACT

High-grade gliomas, including glioblastomas, are malignant brain tumors for which improved treatment is urgently needed. Genetic studies have demonstrated the existence of biologically distinct subsets. Preliminary studies have indicated that platelet-derived growth factor (PDGF) receptor signaling contributes to the growth of some of these tumors. In this study, human high-grade glioma primary cultures were analysed for sensitivity to treatment with the PDGF receptor inhibitor imatinib/Glivec/Gleevec/STI571. Six out of 15 cultures displayed more than 40% growth inhibition after imatinib treatment, whereas seven cultures showed less than 20% growth inhibition. In the sensitive cultures, apoptosis contributed to growth inhibition. Platelet-derived growth factor receptor status correlated with imatinib sensitivity. Supervised analyses of gene expression profiles and real-time PCR analyses identified expression of the chemokine CXCL12/SDF-1 (stromal cell-derived factor 1) as a predictor of imatinib sensitivity. Exogenous addition of CXCL12 to imatinib-insensitive cultures conferred some imatinib sensitivity. Finally, coregulation of CXCL12 and PDGF alpha-receptor was observed in glioblastoma biopsies. We have thus defined the characteristics of a novel imatinib-sensitive subset of glioma cultures, and provided evidence for a functional relationship between imatinib sensitivity and chemokine signaling. These findings will assist in the design and evaluation of clinical trials exploring therapeutic effects of imatinib on malignant brain tumors.


Subject(s)
Antineoplastic Agents/pharmacology , Brain Neoplasms/drug therapy , Glioma/drug therapy , Piperazines/pharmacology , Pyrimidines/pharmacology , Benzamides , Brain Neoplasms/metabolism , Brain Neoplasms/pathology , Chemokine CXCL12 , Chemokines, CXC/biosynthesis , Chemokines, CXC/genetics , Female , Glioma/metabolism , Glioma/pathology , Humans , Imatinib Mesylate , Male , Middle Aged , Receptors, Platelet-Derived Growth Factor/antagonists & inhibitors , Tumor Cells, Cultured
10.
Eur J Cancer ; 40(2): 267-74, 2004 Jan.
Article in English | MEDLINE | ID: mdl-14728942

ABSTRACT

Smad2 and Smad3 are intracellular signal transduction proteins of importance in transforming growth factor-beta (TGFbeta)-mediated inhibition of epithelial cell proliferation. Inactivating mutations in the Smad2 and Smad3 genes have been found in various human malignancies. Here, we show that expression of Smad2 leads to the inhibition of growth of Mv1Lu cells inoculated with Matrigel subcutaneously (s.c.) in severe combined immunodeficient (SCID) mice. In histological appearance, the Matrigel plugs with Smad2-transfected cells showed strongly reduced cell density, proliferation and angiogenesis compared with the small tumour nodules of similar size formed by the vector- or Smad3-transfected cells. The histological appearance of vector- and Smad3-transfected cells inoculated in mice was identical. Overexpression of Smad2 and Smad3 in Mv1Lu cells led to the inhibition of cell growth in three-dimensional cultures when compared with vector-transfected cells. Overexpression of Smad2 and Smad3 also decreased the hyperphosphorylation of pRb in Smad-transfected cells. Thus, increased expression of Smad2 leads to inhibition of Mv1Lu cell proliferation and a reduction in the growth of the Smad2-expressing cells inoculated in mice.


Subject(s)
DNA-Binding Proteins/genetics , Lung Neoplasms/pathology , Trans-Activators/genetics , Animals , Blotting, Western , Gene Expression , Genes, myc/genetics , Immunohistochemistry , Keratinocytes , Mice , Mice, SCID , Neoplasm Transplantation , Phosphorylation , Proteins/genetics , Smad2 Protein , Smad3 Protein , Transfection , Tumor Cells, Cultured
12.
Cancer Res ; 61(15): 5778-83, 2001 Aug 01.
Article in English | MEDLINE | ID: mdl-11479215

ABSTRACT

Dermatofibrosarcoma protuberans (DFSP) and giant cell fibroblastoma (GCF) are recurrent, infiltrative skin tumors that presently are treated with surgery. DFSP and GCF tumors are genetically characterized by chromosomal rearrangements fusing the collagen type Ialpha1 (COLIA1) gene to the platelet-derived growth factor B-chain (PDGFB) gene. It has been shown that the resulting COL1A1/PDGF-B fusion protein is processed to mature PDGF-BB. Autocrine PDGF receptor stimulation has therefore been predicted to contribute to DFSP and GCF tumor development and growth. Here we demonstrate presence of activated PDGF receptors in primary cultures derived from six different DFSP and GCF tumors. Three of the primary cultures were further characterized; their in vitro growth displayed an increased sensitivity to treatment with the PDGF receptor tyrosine kinase inhibitor STI571, as compared with normal fibroblasts. Transplantable tumors, displaying a DFSP-like histology, were established from one of the DFSP primary cultures. Treatment of tumor-bearing severe combined immunodeficient mice with STI571 reduced tumor growth. The growth-inhibitory effects in vitro and in vivo occurred predominantly through induction of tumor cell apoptosis. Our study demonstrates growth-inhibitory effects of PDGF receptor antagonists on human DFSP- and GCF-derived tumor cells and demonstrates that autocrine PDGF receptor stimulation provides antiapoptotic signals contributing to the growth of these cells. These findings suggest targeting of PDGF receptors as a novel treatment strategy for DFSP and GCF.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Dermatofibrosarcoma/pathology , Piperazines/pharmacology , Pyrimidines/pharmacology , Receptor, Platelet-Derived Growth Factor beta/antagonists & inhibitors , Adult , Animals , Benzamides , Cell Division/drug effects , Cell Division/physiology , Child, Preschool , Dermatofibrosarcoma/blood supply , Dermatofibrosarcoma/drug therapy , Female , Fibrosarcoma/blood supply , Fibrosarcoma/drug therapy , Fibrosarcoma/pathology , Giant Cell Tumors/blood supply , Giant Cell Tumors/drug therapy , Giant Cell Tumors/pathology , Growth Inhibitors/pharmacology , Humans , Imatinib Mesylate , Infant , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Mice, SCID , Neovascularization, Pathologic/drug therapy , Oncogene Proteins, Fusion/biosynthesis , Oncogene Proteins, Fusion/genetics , Receptor, Platelet-Derived Growth Factor beta/physiology , Skin Neoplasms/blood supply , Skin Neoplasms/drug therapy , Skin Neoplasms/pathology , Xenograft Model Antitumor Assays
13.
EMBO J ; 20(15): 4132-42, 2001 Aug 01.
Article in English | MEDLINE | ID: mdl-11483516

ABSTRACT

Inhibitory Smads, i.e. Smad6 and Smad7, are potent antagonists of the BMP-Smad pathway by interacting with activated bone morphogenetic protein (BMP) type I receptors and thereby preventing the activation of receptor-regulated Smads, or by competing with activated R-Smads for heteromeric complex formation with Smad4. The molecular mechanisms that underlie the regulation of I-Smad activity have remained elusive. Here we report the identification of a cytoplasmic protein, previously termed associated molecule with the SH3 domain of STAM (AMSH), as a direct binding partner for Smad6. AMSH interacts with Smad6, but not with R- and Co-Smads, upon BMP receptor activation in cultured cells. Consistent with this finding, stimulation of cells with BMP induces a co-localization of Smad6 with AMSH in the cytoplasm. Ectopic expression of AMSH prolongs BMP-induced Smad1 phosphorylation, and potentiates BMP-induced activation of transcriptional reporter activity, growth arrest and apoptosis. The data strongly suggest that the molecular mechanism by which AMSH exerts its action is by inhibiting the binding of Smad6 to activated type I receptors or activated R-Smads.


Subject(s)
Adaptor Proteins, Signal Transducing , Bone Morphogenetic Proteins/metabolism , DNA-Binding Proteins/metabolism , Down-Regulation , Phosphoproteins/metabolism , Signal Transduction , Trans-Activators/metabolism , Transforming Growth Factor beta , Activin Receptors , Animals , Apoptosis , Bone Morphogenetic Protein 7 , COS Cells , Cell Division , Chlorocebus aethiops , Cytoplasm/metabolism , DNA-Binding Proteins/genetics , Endosomal Sorting Complexes Required for Transport , Genes, Reporter , Humans , Luciferases/genetics , Mice , Phosphoproteins/genetics , Phosphorylation , Protein Serine-Threonine Kinases/metabolism , Receptors, Transforming Growth Factor beta/metabolism , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Smad Proteins , Smad1 Protein , Smad4 Protein , Smad5 Protein , Smad6 Protein , Smad7 Protein , Trans-Activators/genetics , Tumor Cells, Cultured
14.
Stem Cells ; 19(4): 295-303, 2001.
Article in English | MEDLINE | ID: mdl-11463949

ABSTRACT

Many aspects of cell behavior, such as growth, motility, differentiation, and apoptosis, are regulated by signals cells receive from their environment. Such signals are important, e.g., during embryonal development, wound healing, hematopoiesis, and in the regulation of the immune response, and may come from interactions with other cells or components of the extracellular matrix, or from binding of soluble signaling molecules to specific receptors at the cell membrane. Hereby different signaling pathways are initiated inside the cell. Perturbations of such signaling pathways are seen in several types of diseases, e.g., cancer, inflammatory conditions, and atherosclerosis. Thus, antagonists of several signaling pathways have potential clinical utility. Several such compounds are currently used or are in clinical trials; others are currently being analyzed in animal models.


Subject(s)
Receptors, Cell Surface/physiology , Signal Transduction , Diabetes Mellitus/drug therapy , Drug Design , Enzyme Inhibitors/therapeutic use , GTP-Binding Proteins/antagonists & inhibitors , GTP-Binding Proteins/metabolism , Humans , Neoplasms/drug therapy , Phosphatidylinositol 3-Kinases/metabolism , Phosphoinositide-3 Kinase Inhibitors , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein Serine-Threonine Kinases/metabolism , Protein-Tyrosine Kinases/antagonists & inhibitors , Protein-Tyrosine Kinases/metabolism
15.
Am J Respir Cell Mol Biol ; 25(1): 60-8, 2001 Jul.
Article in English | MEDLINE | ID: mdl-11472976

ABSTRACT

Changes in the levels of transforming growth factor (TGF)-beta cytokines or receptors observed during the progression of several inflammatory and fibrotic disorders have been used to implicate these cytokines in the pathophysiology of these diseases. Although correlative, these studies were inconclusive because they were unable to demonstrate actual continuous TGF-beta-mediated signaling in the involved tissues. We reasoned that the phosphorylation state and subcellular localization of Smad2, the intracellular effector of TGF-beta/activin-mediated signaling, could be used as a marker of active signaling mediated by these cytokines in situ. We therefore used an experimental model of ovalbumin-induced allergic airway inflammation and were able to demonstrate a dramatic increase in the numbers of bronchial epithelial, alveolar, and infiltrating inflammatory cells expressing nuclear phosphorylated Smad2 within the allergen-challenged lungs. This was accompanied by strong upregulation of the activin receptor ALK-4/ActR-IB and redistribution of the TGF-beta responsive ALK-5/TbetaR-I. Although levels of TGF-beta1, TGF-beta2, and TGF-beta3 messenger RNA (mRNA) were marginally altered, the level of activin mRNA was strongly upregulated during the inflammatory response. Our data illustrate the usefulness of antiphosphorylated Smad antibodies in demonstrating active TGF- beta/activin-mediated signaling in vivo and strongly suggest that activin/Smad-mediated signaling could be a critical contributor in the pathophysiology of allergic pulmonary diseases.


Subject(s)
Bronchitis/metabolism , DNA-Binding Proteins/metabolism , Hypersensitivity/metabolism , Inhibins/metabolism , Trans-Activators/metabolism , Transforming Growth Factor beta/metabolism , Activins , Animals , Base Sequence , Blotting, Western , Bronchitis/etiology , DNA Primers , Female , Hypersensitivity/complications , Immunohistochemistry , Mice , Mice, Inbred BALB C , Phosphorylation , Protein Transport , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction , Smad2 Protein
17.
J Biol Chem ; 276(30): 27749-52, 2001 Jul 27.
Article in English | MEDLINE | ID: mdl-11390370

ABSTRACT

Ligand binding to the platelet-derived growth factor (PDGF) beta-receptor leads to increased receptor tyrosine phosphorylation as a consequence of dimerization-induced activation of the intrinsic receptor tyrosine kinase activity. In this study we asked whether ligand-stimulated PDGF beta-receptor tyrosine phosphorylation, to some extent, also involved reduced susceptibility to tyrosine dephosphorylation. To investigate this possibility we compared the sensitivity of ligand-stimulated and non-stimulated forms of tyrosine-phosphorylated PDGF beta-receptors to dephosphorylation using various preparations containing protein-tyrosine phosphatase activity. Ligand-stimulated or unstimulated tyrosine-phosphorylated receptors were obtained after incubation of cells with pervanadate only or pervanadate, together with PDGF-BB, respectively. Dephosphorylation of receptors immobilized on wheat germ agglutinin-Sepharose, as well as of receptors in intact cell membranes, was investigated under conditions when rephosphorylation did not occur. As compared with unstimulated receptors the ligand-stimulated PDGF beta-receptors showed about 10-fold reduced sensitivity to dephosphorylation by cell membranes, a recombinant form of the catalytic domain of density-enhanced phosphatase-1, or recombinant protein-tyrosine phosphatase 1B. We conclude that ligand-stimulated forms of the PDGF beta-receptor display a reduced susceptibility to dephosphorylation. Our findings suggest a novel mechanism whereby ligand stimulation of PDGF beta-receptor, and possibly other tyrosine kinase receptors, leads to a net increase in receptor tyrosine phosphorylation.


Subject(s)
Receptor, Platelet-Derived Growth Factor beta/metabolism , Tyrosine/metabolism , Agglutinins/metabolism , Animals , Catalytic Domain , Cell Membrane/enzymology , Cell Membrane/metabolism , Cells, Cultured , Dimerization , Endothelium, Vascular/cytology , Enzyme Inhibitors/pharmacology , Ligands , Phosphorylation , Precipitin Tests , Protein Binding , Protein Phosphatase 1 , Protein Structure, Tertiary , Protein Tyrosine Phosphatases/metabolism , Recombinant Proteins/metabolism , Sepharose/chemistry , Sepharose/metabolism , Swine , Transfection , Vanadates/pharmacology
18.
Nat Cell Biol ; 3(5): 512-6, 2001 May.
Article in English | MEDLINE | ID: mdl-11331881

ABSTRACT

The term 'platelet-derived growth factor' (PDGF) refers to a family of disulphide-bonded dimeric isoforms that are important for growth, survival and function in several types of connective tissue cell. So far, three different PDGF chains have been identified - the classical PDGF-A and PDGF-B and the recently identified PDGF-C. PDGF isoforms (PDGF-AA, AB, BB and CC) exert their cellular effects by differential binding to two receptor tyrosine kinases. The PDGF alpha-receptor (PDGFR-alpha) binds to all three PDGF chains, whereas the beta-receptor (PDGFR-beta) binds only to PDGF-B. Gene-targeting studies using mice have shown that the genes for PDGF-A and PDGF-B, as well as the two PDGFR genes, are essential for normal development. Furthermore, overexpression of PDGFs is linked to different pathological conditions, including malignancies, atherosclerosis and fibroproliferative diseases. Here we have identify and characterize a fourth member of the PDGF family, PDGF-D. PDGF-D has a two-domain structure similar to PDGF-C and is secreted as a disulphide-linked homodimer, PDGF-DD. Upon limited proteolysis, PDGF-DD is activated and becomes a specific agonistic ligand for PDGFR-beta. PDGF-DD is the first known PDGFR-beta-specific ligand, and its unique receptor specificity indicates that it may be important for development and pathophysiology in several organs.


Subject(s)
Lymphokines , Platelet-Derived Growth Factor/chemistry , Receptor, Platelet-Derived Growth Factor beta/chemistry , Amino Acid Sequence , Animals , Baculoviridae/metabolism , Blotting, Northern , Cloning, Molecular , Cysteine/chemistry , DNA, Complementary/metabolism , Dimerization , Dose-Response Relationship, Drug , Humans , Immunohistochemistry , Insecta , Ligands , Mice , Mice, Transgenic , Molecular Sequence Data , Phylogeny , Protein Binding , Protein Structure, Tertiary , RNA, Messenger/metabolism , Receptor, Platelet-Derived Growth Factor beta/metabolism , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Sequence Homology, Amino Acid , Tissue Distribution
19.
Mol Biol Cell ; 12(4): 1079-91, 2001 Apr.
Article in English | MEDLINE | ID: mdl-11294908

ABSTRACT

Smad proteins are cytoplasmic signaling effectors of transforming growth factor-beta (TGF-beta) family cytokines and regulate gene transcription in the nucleus. Receptor-activated Smads (R-Smads) become phosphorylated by the TGF-beta type I receptor. Rapid and precise transport of R-Smads to the nucleus is of crucial importance for signal transduction. By focusing on the R-Smad Smad3 we demonstrate that 1) only activated Smad3 efficiently enters the nucleus of permeabilized cells in an energy- and cytosol-dependent manner. 2) Smad3, via its N-terminal domain, interacts specifically with importin-beta1 and only after activation by receptor. In contrast, the unique insert of exon3 in the N-terminal domain of Smad2 prevents its association with importin-beta1. 3) Nuclear import of Smad3 in vivo requires the action of the Ran GTPase, which mediates release of Smad3 from the complex with importin-beta1. 4) Importin-beta1, Ran, and p10/NTF2 are sufficient to mediate import of activated Smad3. The data describe a pathway whereby Smad3 phosphorylation by the TGF-beta receptor leads to enhanced interaction with importin-beta1 and Ran-dependent import and release into the nucleus. The import mechanism of Smad3 shows distinct features from that of the related Smad2 and the structural basis for this difference maps to the divergent sequences of their N-terminal domains.


Subject(s)
Cell Nucleus/metabolism , DNA-Binding Proteins/metabolism , Nuclear Proteins/metabolism , Nucleocytoplasmic Transport Proteins , Trans-Activators/metabolism , Transforming Growth Factor beta/metabolism , ran GTP-Binding Protein/metabolism , Active Transport, Cell Nucleus , Animals , Binding Sites , Carrier Proteins/metabolism , Cell Line , Cell Line, Transformed , Humans , Karyopherins , Mice , Signal Transduction , Smad2 Protein , Smad3 Protein , Tumor Cells, Cultured
20.
Cancer Res ; 61(7): 2929-34, 2001 Apr 01.
Article in English | MEDLINE | ID: mdl-11306470

ABSTRACT

Most solid malignancies display interstitial hypertension and a poor uptake of anticancer drugs. Platelet-derived growth factor (PDGF) and the cognate tyrosine kinase receptors are expressed in many tumors. Signaling through PDGFbeta receptors was shown recently to increase interstitial fluid pressure (IFP) in dermis after anaphylaxis-induced lowering of IFP. In this study, we show that treatment with the selective PDGF receptor kinase inhibitor, STI571, formerly known as CGP57148B, decreased the interstitial hypertension and increased capillary-to-interstitium transport of 51Cr-EDTA in s.c. growing rat PROb colonic carcinomas. Furthermore, treatment with an antagonistic PDGF-B oligonucleotide aptamer decreased interstitial hypertension in these tumors. PDGFbeta receptors were expressed in blood vessels and stromal cells but not in the tumor cells of PROb colonic carcinomas. Our study indicates a previously unrecognized role of PDGF receptors in tumor biology, although similar effects of PDGF on IFP have been demonstrated previously in the dermis. The data suggest interference with PDGF receptors, or their ligands, as a novel strategy to increase drug uptake and therapeutic effectiveness of cancer chemotherapy.


Subject(s)
Colonic Neoplasms/metabolism , Receptors, Platelet-Derived Growth Factor/antagonists & inhibitors , Animals , Antineoplastic Agents/pharmacology , Benzamides , Capillary Permeability/drug effects , Capillary Permeability/physiology , Colonic Neoplasms/blood supply , Edetic Acid/pharmacokinetics , Enzyme Inhibitors/pharmacology , Extracellular Matrix Proteins/metabolism , Imatinib Mesylate , Microdialysis , Oligonucleotides/pharmacology , Phosphorylation/drug effects , Piperazines/pharmacology , Proto-Oncogene Proteins/metabolism , Pyrimidines/pharmacology , Rats , Receptor Protein-Tyrosine Kinases/metabolism , Receptor, Platelet-Derived Growth Factor beta/metabolism , Substrate Specificity , Vascular Endothelial Growth Factor Receptor-1
SELECTION OF CITATIONS
SEARCH DETAIL